1
|
Pourzangiabadi M, Najafi H, Fallah A, Goudarzi A, Pouladi I. Dengue virus: Etiology, epidemiology, pathobiology, and developments in diagnosis and control - A comprehensive review. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2025; 127:105710. [PMID: 39732271 DOI: 10.1016/j.meegid.2024.105710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Dengue flavivirus (DENV) is the virus that causes dengue, one of the most dangerous and common viral diseases in humans that are carried by mosquitoes and can lead to fatalities. Every year, there are over 400 million cases of dengue fever worldwide, and 22,000 fatalities. It has been documented in tropical and subtropical climates in over 100 nations. Unfortunately, there is no specific treatment approach, but prevention, adequate awareness, diagnosis in the early stages of viral infection and proper medical care can reduce the mortality rate. The first licensed vaccine for dengue virus (CYD Denvaxia) was quadrivalent, but it is not approved in all countries. The primary barriers to vaccine development include inadequate animal models, inadequate etiology mechanistic studies, and adverse drug events. This study provides current knowledge and a comprehensive view of the biology, production and reproduction, transmission, pathogenesis and diagnosis, epidemiology and control measures of dengue virus.
Collapse
Affiliation(s)
- Masoud Pourzangiabadi
- Department of Microbiology, Faculty of Science, Kerman Branch, Islamic Azad University, Kerman, Iran
| | - Hamideh Najafi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Arezoo Fallah
- Department of Bacteriology and Virology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Aida Goudarzi
- Department of Clinical Science, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Iman Pouladi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
2
|
Saihar A, Yaseen AR, Suleman M, Parveen R, Bashir H. From bytes to bites: In-silico creation of a novel multi-epitope vaccine against Murray Valley Encephalitis Virus. Microb Pathog 2025; 198:107171. [PMID: 39617074 DOI: 10.1016/j.micpath.2024.107171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/15/2024]
Abstract
Flaviviruses transmitted by arthropods, including the Murray Valley Encephalitis Virus (MVEV), are RNA viruses capable of causing severe encephalitis in various hosts. The spread of these viruses is closely linked to climatic conditions and the habitats of host and vector species, leading to outbreaks in new geographic regions. Notable encephalitis-causing flaviviruses include Japanese encephalitis virus (JEV), West Nile virus (WNV), and Kunjin virus (KUNV). MVEV, primarily spread by the mosquito Culex annulirostris and amplified by water birds such as egrets and Nankeen night herons, has caused significant outbreaks in Australia, including severe epidemics in 1951, 1956, and 1974. Despite its severity, no rapid diagnostic techniques or effective antiviral treatments are available, and current interventions are limited to supportive care and mosquito management. Given the absence of a licensed vaccine, this study aimed to develop a multi-epitope hybrid vaccine targeting MVEV using in silico approaches. The study focused on identifying B-cell and T-cell epitopes from the MVEV Envelope (E) protein, constructing a vaccine candidate, and computationally validating its immunogenic potential. The designed vaccine underwent rigorous analysis of its antigenic properties, allergenicity, and toxicity. Disulfide engineering and assessment of physicochemical properties ensured the structural integrity of the vaccine, supported by Ramachandran plot and ProSA web analyses. Molecular docking studies assessed the vaccine's binding affinities with TLR-3, and MHC-I. Population coverage analysis of MHC-I and MHC-II epitopes evaluated global efficacy. Additionally, molecular dynamics simulations explored the stability of docked complexes, and PDBsum analysis elucidated interaction details. Immunological simulations were conducted to predict immune response outcomes, providing comprehensive validation of the vaccine's antigenicity. The findings highlight the potential of a multi-epitope vaccine as a viable strategy for MVEV prevention.
Collapse
Affiliation(s)
- Aisha Saihar
- Center for Applied Molecular Biology, CAMB, University of the Punjab, Lahore, Pakistan.
| | - Allah Rakha Yaseen
- School of Biological Sciences, University of the Punjab, Lahore 54590, Pakistan.
| | - Muhammad Suleman
- School of Biological Sciences, University of the Punjab, Lahore 54590, Pakistan.
| | - Rukhsana Parveen
- Center for Applied Molecular Biology, CAMB, University of the Punjab, Lahore, Pakistan.
| | - Hamid Bashir
- Center for Applied Molecular Biology, CAMB, University of the Punjab, Lahore, Pakistan.
| |
Collapse
|
3
|
Cavina L, Bouma MJ, Gironés D, Feiters MC. Orthoflaviviral Inhibitors in Clinical Trials, Preclinical In Vivo Efficacy Targeting NS2B-NS3 and Cellular Antiviral Activity via Competitive Protease Inhibition. Molecules 2024; 29:4047. [PMID: 39274895 PMCID: PMC11396989 DOI: 10.3390/molecules29174047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/16/2024] Open
Abstract
Orthoflaviviruses, including zika (ZIKV), West Nile (WNV), and dengue (DENV) virus, induce severely debilitating infections and contribute significantly to the global disease burden, yet no clinically approved antiviral treatments exist. This review offers a comprehensive analysis of small-molecule drug development targeting orthoflaviviral infections, with a focus on NS2B-NS3 inhibition. We systematically examined clinical trials, preclinical efficacy studies, and modes of action for various viral replication inhibitors, emphasizing allosteric and orthosteric drugs inhibiting NS2B-NS3 protease with in vivo efficacy and in vitro-tested competitive NS2B-NS3 inhibitors with cellular efficacy. Our findings revealed that several compounds with in vivo preclinical efficacy failed to show clinical antiviral efficacy. NS3-NS4B inhibitors, such as JNJ-64281802 and EYU688, show promise, recently entering clinical trials, underscoring the importance of developing novel viral replication inhibitors targeting viral machinery. To date, the only NS2B-NS3 inhibitor that has undergone clinical trials is doxycycline, however, its mechanism of action and clinical efficacy as viral growth inhibitor require additional investigation. SYC-1307, an allosteric inhibitor, exhibits high in vivo efficacy, while temoporfin and methylene blue represent promising orthosteric non-competitive inhibitors. Compound 71, a competitive NS2B-NS3 inhibitor, emerges as a leading preclinical candidate due to its high cellular antiviral efficacy, minimal cytotoxicity, and favorable in vitro pharmacokinetic parameters. Challenges remain in developing competitive NS2B-NS3 inhibitors, including appropriate biochemical inhibition assays as well as the selectivity and conformational flexibility of the protease, complicating effective antiviral treatment design.
Collapse
Affiliation(s)
- Lorenzo Cavina
- Institute for Molecules and Materials, Faculty of Science, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (M.J.B.); (D.G.)
| | - Mathijs J. Bouma
- Institute for Molecules and Materials, Faculty of Science, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (M.J.B.); (D.G.)
| | - Daniel Gironés
- Institute for Molecules and Materials, Faculty of Science, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (M.J.B.); (D.G.)
- Protinhi Therapeutics, Transistorweg 5, 6534 AT Nijmegen, The Netherlands
| | - Martin C. Feiters
- Institute for Molecules and Materials, Faculty of Science, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; (M.J.B.); (D.G.)
| |
Collapse
|
4
|
Wang ZJ, Zhang RR, Wu M, Zhao H, Li XF, Ye Q, Qin CF. Development of a live-attenuated chimeric vaccine against the emerging Usutu virus. Vaccine 2024; 42:1363-1371. [PMID: 38310016 DOI: 10.1016/j.vaccine.2024.01.077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/30/2023] [Accepted: 01/23/2024] [Indexed: 02/05/2024]
Abstract
Usutu virus (USUV) is an emerging arthropod-borne flavivirus that has expanded into multiple European countries during the past several decades. USUV infection in human has been linked to severe neurological complications, and no vaccine is now available against USUV. In this work, we develop a live-attenuated chimeric USUV vaccine (termed ChinUSUV) based on the full-length infectious cDNA clone of the licensed Japanese encephalitis virus (JEV) vaccine strain SA14-14-2. In vitro studies demonstrate that ChinUSUV replicates efficiently and maintains its genetic stability. Remarkably, ChinUSUV exhibits a significant attenuation phenotype in multiple mouse models even compared with the licensed JEV vaccine. A single immunization with ChinUSUV elicits potent IgG and neutralizing antibody responses as well as T cell response. Passive transfer of sera from ChinUSUV-immunized mice confers significant protection against lethal homologous challenge in suckling mice. Taken together, our results suggest that ChinUSUV represents a potential USUV vaccine candidate that merits further development.
Collapse
Affiliation(s)
- Zheng-Jian Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Rong-Rong Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Mei Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Hui Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Xiao-Feng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Qing Ye
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing 100071, China.
| |
Collapse
|
5
|
da Silva Rodrigues JV, Rodrigues Gazolla PA, da Cruz Pereira I, Dias RS, Poly da Silva IE, Oliveira Prates JW, de Souza Gomes I, de Azevedo Silveira S, Costa AV, de Oliveira FM, de Aguiar AR, Canedo da Silva C, Teixeira RR, de Paula SO. Synthesis and virucide activity on zika virus of 1,2,3-triazole-containing vanillin derivatives. Antiviral Res 2023; 212:105578. [PMID: 36934985 DOI: 10.1016/j.antiviral.2023.105578] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/19/2023]
Abstract
The Zika virus (ZIKV) is an arbovirus and belongs to the Flaviviridae family and Flavivirus genus, with dissemination in the Americas. In Brazil, the predominant strain is the Asian, promoting outbreaks that started in 2015 and are directly related to microcephaly in newborns and Guillain-Barré syndrome in adults. Recently, researchers identified a new African strain circulating in Brazil at the mid-end of 2018 and the beginning of 2019, with the potential to originate a new epidemic. To date, there is no approved vaccine or drug for the treatment of Zika syndrome, and the development of therapeutic alternatives to treat it is of relevance. A critical approach is to use natural products when searching for new chemical agents to treat Zika syndrome. The present investigation describes the preparation of a series of 1,2,3-triazoles derived from the natural product vanillin and the evaluation of their virucide activity. A series of fourteen derivatives were prepared via alkylation of vanillin followed by CuAAC (the copper(I)-catalyzed azide-alkyne cycloaddition) reaction. The compounds were fully characterized by infrared (I.R.), nuclear magnetic resonance (NMR), and high-resolution mass spectrometry (HRMS) techniques. The cytotoxicity of Vero cells and the effect on the Zika Virus of the vanillin derivatives were evaluated. It was found that the most effective compound corresponded to 4-((1-(4-isopropylbenzyl)-1H-1,2,3-triazol-4-yl)methoxy)-3-methoxybenzaldehyde (8) (EC50 = 27.14 μM, IC50 = 334.9 μM). Subsequent assessments, namely pre and post-treatment assays, internalization and adsorption inhibition assays, kinetic, electronic microscopy analyses, and zeta potential determination, revealed that compound 8 blocks the Zika virus infection in vitro by acting on the viral particle. A molecular docking study was performed, and the results are also discussed.
Collapse
Affiliation(s)
- João Vitor da Silva Rodrigues
- Laboratório de Imunovirologia Molecular, Departamento de Biologia Geral, Universidade Federal de Viçosa, Minas Gerais, Brazil
| | | | - Iago da Cruz Pereira
- Laboratório de Imunovirologia Molecular, Departamento de Biologia Geral, Universidade Federal de Viçosa, Minas Gerais, Brazil
| | - Roberto Sousa Dias
- Laboratório de Imunovirologia Molecular, Departamento de Biologia Geral, Universidade Federal de Viçosa, Minas Gerais, Brazil
| | | | - John Willians Oliveira Prates
- Laboratório de Imunovirologia Molecular, Departamento de Biologia Geral, Universidade Federal de Viçosa, Minas Gerais, Brazil
| | - Isabela de Souza Gomes
- Departamento de Ciência da Computação, Universidade Federal de Viçosa, Minas Gerais, Brazil
| | | | - Adilson Vidal Costa
- Departamento de Química e Física, Universidade Federal do Espírito Santo, Espírito Santo, Brazil
| | | | | | | | | | - Sérgio Oliveira de Paula
- Laboratório de Imunovirologia Molecular, Departamento de Biologia Geral, Universidade Federal de Viçosa, Minas Gerais, Brazil.
| |
Collapse
|
6
|
Liu X, Chen X, Dong Y, Zhang C, Qu X, Lei Y, Jiang Z, Wei X. Multiple virus sorting based on aptamer-modified microspheres in a TSAW device. MICROSYSTEMS & NANOENGINEERING 2023; 9:64. [PMID: 37213822 PMCID: PMC10192341 DOI: 10.1038/s41378-023-00523-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/15/2023] [Accepted: 03/19/2023] [Indexed: 05/23/2023]
Abstract
Due to the overlapping epidemiology and clinical manifestations of flaviviruses, differential diagnosis of these viral diseases is complicated, and the results are unreliable. There is perpetual demand for a simplified, sensitive, rapid and inexpensive assay with less cross-reactivity. The ability to sort distinct virus particles from a mixture of biological samples is crucial for improving the sensitivity of diagnoses. Therefore, we developed a sorting system for the subsequent differential diagnosis of dengue and tick-borne encephalitis in the early stage. We employed aptamer-modified polystyrene (PS) microspheres with different diameters to specifically capture dengue virus (DENV) and tick-borne encephalitis virus (TBEV), and utilized a traveling surface acoustic wave (TSAW) device to accomplish microsphere sorting according to particle size. The captured viruses were then characterized by laser scanning confocal microscopy (LSCM), field emission scanning electron microscopy (FE-SEM) and reverse transcription-polymerase chain reaction (RT‒PCR). The characterization results indicated that the acoustic sorting process was effective and damage-free for subsequent analysis. Furthermore, the strategy can be utilized for sample pretreatment in the differential diagnosis of viral diseases.
Collapse
Affiliation(s)
- Xianglian Liu
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an, 710049 China
| | - Xuan Chen
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an, 710049 China
| | - Yangchao Dong
- Department of Microbiology, the Fourth Military Medical University, Xi’an, 710032 China
| | - Chuanyu Zhang
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an, 710049 China
| | - Xiaoli Qu
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an, 710049 China
| | - Yingfeng Lei
- Department of Microbiology, the Fourth Military Medical University, Xi’an, 710032 China
| | - Zhuangde Jiang
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an, 710049 China
| | - Xueyong Wei
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an, 710049 China
| |
Collapse
|
7
|
Samrat SK, Xu J, Li Z, Zhou J, Li H. Antiviral Agents against Flavivirus Protease: Prospect and Future Direction. Pathogens 2022; 11:293. [PMID: 35335617 PMCID: PMC8955721 DOI: 10.3390/pathogens11030293] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/15/2022] [Accepted: 02/20/2022] [Indexed: 12/18/2022] Open
Abstract
Flaviviruses cause a significant amount of mortality and morbidity, especially in regions where they are endemic. A recent example is the outbreak of Zika virus throughout the world. Development of antiviral drugs against different viral targets is as important as the development of vaccines. During viral replication, a single polyprotein precursor (PP) is produced and further cleaved into individual proteins by a viral NS2B-NS3 protease complex together with host proteases. Flavivirus protease is one of the most attractive targets for development of therapeutic antivirals because it is essential for viral PP processing, leading to active viral proteins. In this review, we have summarized recent development in drug discovery targeting the NS2B-NS3 protease of flaviviruses, especially Zika, dengue, and West Nile viruses.
Collapse
Affiliation(s)
- Subodh K. Samrat
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (S.K.S.); (Z.L.)
| | - Jimin Xu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA; (J.X.); (J.Z.)
| | - Zhong Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (S.K.S.); (Z.L.)
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA; (J.X.); (J.Z.)
| | - Hongmin Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1703 E Mabel St, Tucson, AZ 85721, USA; (S.K.S.); (Z.L.)
- BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
8
|
Abstract
Mass spectrometry imaging (MSI) is a label-free molecular imaging technique allowing an untargeted detection of a broad range of biomolecules and xenobiotics. MSI enables imaging of the spatial distribution of proteins, peptides, lipids and metabolites from a wide range of samples. To date, this technique is commonly applied to tissue sections in cancer diagnostics and biomarker development, but also molecular histology in general. Advances in the methodology and bioinformatics improved the resolution of MS images below the single cell level and increased the flexibility of the workflow. However, MSI-based research in virology is just starting to gain momentum and its full potential has not been exploited yet. In this review, we discuss the main applications of MSI in virology. We review important aspects of matrix-assisted laser desorption/ionization (MALDI) MSI, the most widely used MSI technique in virology. In addition, we summarize relevant literature on MSI studies that aim to unravel virus-host interactions and virus pathogenesis, to elucidate antiviral drug kinetics and to improve current viral disease diagnostics. Collectively, these studies strongly improve our general understanding of virus-induced changes in the proteome, metabolome and metabolite distribution in host tissues of humans, animals and plants upon infection. Furthermore, latest MSI research provided important insights into the drug distribution and distribution kinetics, especially in antiretroviral research. Finally, MSI-based investigations of oncogenic viruses greatly increased our knowledge on tumor mass signatures and facilitated the identification of cancer biomarkers.
Collapse
Affiliation(s)
- Luca D Bertzbach
- Institute of Virology, Freie Universität Berlin, Berlin, Germany
| | | | - Axel Karger
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany.
| |
Collapse
|
9
|
Xisto MF, Dias RS, Feitosa-Araujo E, Prates JWO, da Silva CC, de Paula SO. Efficient Plant Production of Recombinant NS1 Protein for Diagnosis of Dengue. FRONTIERS IN PLANT SCIENCE 2020; 11:581100. [PMID: 33193526 PMCID: PMC7649140 DOI: 10.3389/fpls.2020.581100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/02/2020] [Indexed: 05/28/2023]
Abstract
Dengue fever is endemic in more than 120 countries, which account for 3.9 billion people at risk of infection worldwide. The absence of a vaccine with effective protection against the four serotypes of this virus makes differential molecular diagnosis the key step for the correct treatment of the disease. Rapid and efficient diagnosis prevents progression to a more severe stage of this disease. Currently, the limiting factor in the manufacture of dengue (DENV) diagnostic kits is the lack of large-scale production of the non-structural 1 (NS1) protein (antigen) to be used in the capture of antibodies from the blood serum of infected patients. In this work, we use plant biotechnology and genetic engineering as tools for the study of protein production for research and commercial purposes. Gene transfer, integration and expression in plants is a valid strategy for obtaining large-scale and low-cost heterologous protein production. The authors produced NS1 protein of the dengue virus serotype 2 (NS1DENV2) in the Arabidopsis thaliana plant. Transgenic plants obtained by genetic transformation expressed the recombinant protein that was purified and characterized for diagnostic use. The yield was 203 μg of the recombinant protein per gram of fresh leaf. By in situ immunolocalization, transgenic protein was observed within the plant tissue, located in aggregates bodies. These antigens showed high sensitivity and specificity to both IgM (84.29% and 91.43%, respectively) and IgG (83.08% and 87.69%, respectively). The study goes a step further to validate the use of plants as a strategy for obtaining large-scale and efficient protein production to be used in dengue virus diagnostic tests.
Collapse
Affiliation(s)
| | - Roberto Sousa Dias
- Department of General Biology, Federal University of Viçosa, Viçosa, Brazil
| | | | | | | | | |
Collapse
|
10
|
The Molecular Interactions of ZIKV and DENV with the Type-I IFN Response. Vaccines (Basel) 2020; 8:vaccines8030530. [PMID: 32937990 PMCID: PMC7565347 DOI: 10.3390/vaccines8030530] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022] Open
Abstract
Zika Virus (ZIKV) and Dengue Virus (DENV) are related viruses of the Flavivirus genus that cause significant disease in humans. Existing control measures have been ineffective at curbing the increasing global incidence of infection for both viruses and they are therefore prime targets for new vaccination strategies. Type-I interferon (IFN) responses are important in clearing viral infection and for generating efficient adaptive immune responses towards infection and vaccination. However, ZIKV and DENV have evolved multiple molecular mechanisms to evade type-I IFN production. This review covers the molecular interactions, from detection to evasion, of these viruses with the type-I IFN response. Additionally, we discuss how this knowledge can be exploited to improve the design of new vaccine strategies.
Collapse
|
11
|
West Nile Virus: An Update on Pathobiology, Epidemiology, Diagnostics, Control and "One Health" Implications. Pathogens 2020; 9:pathogens9070589. [PMID: 32707644 PMCID: PMC7400489 DOI: 10.3390/pathogens9070589] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/16/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023] Open
Abstract
West Nile virus (WNV) is an important zoonotic flavivirus responsible for mild fever to severe, lethal neuroinvasive disease in humans, horses, birds, and other wildlife species. Since its discovery, WNV has caused multiple human and animal disease outbreaks in all continents, except Antarctica. Infections are associated with economic losses, mainly due to the cost of treatment of infected patients, control programmes, and loss of animals and animal products. The pathogenesis of WNV has been extensively investigated in natural hosts as well as in several animal models, including rodents, lagomorphs, birds, and reptiles. However, most of the proposed pathogenesis hypotheses remain contentious, and much remains to be elucidated. At the same time, the unavailability of specific antiviral treatment or effective and safe vaccines contribute to the perpetuation of the disease and regular occurrence of outbreaks in both endemic and non-endemic areas. Moreover, globalisation and climate change are also important drivers of the emergence and re-emergence of the virus and disease. Here, we give an update of the pathobiology, epidemiology, diagnostics, control, and “One Health” implications of WNV infection and disease.
Collapse
|
12
|
Abstract
Flaviviruses are a genus of mostly arthropod-borne RNA viruses that cause a range of pathologies in humans. Basic knowledge on flaviviruses is rapidly expanding, partly due to their status as frequent emerging or re-emerging pathogens. Flaviviruses include the dengue, Zika, West Nile, tick-borne encephalitis and yellow fever viruses (DENV, ZIKV, WNV, TBEV and YFV, respectively). As is the case with other families of viruses, the success of productive infection of human cells by flaviviruses depends in part on the antiviral activity of a heterogeneous group of cellular antiviral proteins called restriction factors. Restriction factors are the effector proteins of the cell-autonomous innate response against viruses, an immune pathway that also includes virus sensors as well as intracellular and extracellular signal mediators such as type I interferons (IFN-I). In this review, I summarize recent progress toward the identification and characterization of flavivirus restriction factors. In particular, I focus on IFI6, Schlafen 11, FMRP, OAS-RNase L, RyDEN, members of the TRIM family of proteins (TRIM5α, TRIM19, TRIM56, TRIM69 and TRIM79α) and a new mechanism of action proposed for viperin. Recent and future studies on this topic will lead to a more complete picture of the flavivirus restrictome, defined as the ensemble of cellular factors with demonstrated anti-flaviviral activity.
Collapse
|
13
|
Revisiting Key Entry Routes of Human Epidemic Arboviruses into the Mainland Americas through Large-Scale Phylogenomics. Int J Genomics 2018; 2018:6941735. [PMID: 30402454 PMCID: PMC6196792 DOI: 10.1155/2018/6941735] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/24/2018] [Accepted: 08/02/2018] [Indexed: 11/18/2022] Open
Abstract
The rapid worldwide spread of chikungunya (CHIKV), dengue (DENV), and Zika (ZIKV) viruses have raised great international concern. Knowledge about the entry routes and geographic expansion of these arboviruses to the mainland Americas remain incomplete and controversial. Epidemics caused by arboviruses continue to cause socioeconomic burden globally, particularly in countries where vector control is difficult due to climatic or infrastructure factors. Understanding how the virus circulates and moves from one country to another is of paramount importance to assist government and health officials in anticipating future epidemics, as well as to take steps to help control or mitigate the spread of the virus. Through the analyses of the sequences of arbovirus genomes collected at different locations over time, we identified patterns of accumulated mutations, being able to trace routes of dispersion of these viruses. Here, we applied robust phylogenomic methods to trace the evolutionary dynamics of these arboviruses with special focus on Brazil, the epicenter of these triple epidemics. Our results show that CHIKV, DENV-1–4, and ZIKV followed a similar path prior to their first introductions into the mainland Americas, underscoring the need for systematic arboviral surveillance at major entry points of human population movement between countries such as airports and seaports.
Collapse
|
14
|
Zheng Z, Yang J, Jiang X, Liu Y, Zhang X, Li M, Zhang M, Fu M, Hu K, Wang H, Luo MH, Gong P, Hu Q. Tick-Borne Encephalitis Virus Nonstructural Protein NS5 Induces RANTES Expression Dependent on the RNA-Dependent RNA Polymerase Activity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:53-68. [PMID: 29760190 DOI: 10.4049/jimmunol.1701507] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 04/30/2018] [Indexed: 01/08/2023]
Abstract
Tick-borne encephalitis virus (TBEV) is one of the flaviviruses that targets the CNS and causes encephalitis in humans. The mechanism of TBEV that causes CNS destruction remains unclear. It has been reported that RANTES-mediated migration of human blood monocytes and T lymphocytes is specifically induced in the brain of mice infected with TBEV, which causes ensuing neuroinflammation and may contribute to brain destruction. However, the viral components responsible for RANTES induction and the underlying mechanisms remain to be fully addressed. In this study, we demonstrate that the NS5, but not other viral proteins of TBEV, induces RANTES production in human glioblastoma cell lines and primary astrocytes. TBEV NS5 appears to activate the IFN regulatory factor 3 (IRF-3) signaling pathway in a manner dependent on RIG-I/MDA5, which leads to the nuclear translocation of IRF-3 to bind with RANTES promoter. Further studies reveal that the activity of RNA-dependent RNA polymerase (RdRP) but not the RNA cap methyltransferase is critical for TBEV NS5-induced RANTES expression, and this is likely due to RdRP-mediated synthesis of dsRNA. Additional data indicate that the residues at K359, D361, and D664 of TBEV NS5 are critical for RdRP activity and RANTES induction. Of note, NS5s from other flaviviruses, including Japanese encephalitis virus, West Nile virus, Zika virus, and dengue virus, can also induce RANTES expression, suggesting the significance of NS5-induced RANTES expression in flavivirus pathogenesis. Our findings provide a foundation for further understanding how flaviviruses cause neuroinflammation and a potential viral target for intervention.
Collapse
Affiliation(s)
- Zifeng Zheng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jieyu Yang
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xuan Jiang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yalan Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China;
| | - Xiaowei Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Mei Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mudan Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou 510623, China; and
| | - Ming Fu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kai Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Hanzhong Wang
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Min-Hua Luo
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peng Gong
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Qinxue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China;
- Institute for Infection and Immunity, St George's, University of London, London SW17 0RE, United Kingdom
| |
Collapse
|
15
|
Stabell AC, Meyerson NR, Gullberg RC, Gilchrist AR, Webb KJ, Old WM, Perera R, Sawyer SL. Dengue viruses cleave STING in humans but not in nonhuman primates, their presumed natural reservoir. eLife 2018; 7:31919. [PMID: 29557779 PMCID: PMC5860865 DOI: 10.7554/elife.31919] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 02/08/2018] [Indexed: 12/25/2022] Open
Abstract
Human dengue viruses emerged from primate reservoirs, yet paradoxically dengue does not reach high titers in primate models. This presents a unique opportunity to examine the genetics of spillover versus reservoir hosts. The dengue virus 2 (DENV2) - encoded protease cleaves human STING, reducing type I interferon production and boosting viral titers in humans. We find that both human and sylvatic (reservoir) dengue viruses universally cleave human STING, but not the STING of primates implicated as reservoir species. The special ability of dengue to cleave STING is thus specific to humans and a few closely related ape species. Conversion of residues 78/79 to the human-encoded 'RG' renders all primate (and mouse) STINGs sensitive to viral cleavage. Dengue viruses may have evolved to increase viral titers in the dense and vast human population, while maintaining decreased titers and pathogenicity in the more rare animals that serve as their sustaining reservoir in nature.
Collapse
Affiliation(s)
- Alex C Stabell
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
| | - Nicholas R Meyerson
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
| | - Rebekah C Gullberg
- Arthropod-borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, United States
| | - Alison R Gilchrist
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
| | - Kristofor J Webb
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
| | - William M Old
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
| | - Rushika Perera
- Arthropod-borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, United States
| | - Sara L Sawyer
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
| |
Collapse
|
16
|
Ncube NB, Ramharack P, Soliman MES. An “All-In-One” Pharmacophoric Architecture for the Discovery of Potential Broad-Spectrum Anti-Flavivirus Drugs. Appl Biochem Biotechnol 2018; 185:799-814. [DOI: 10.1007/s12010-017-2690-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 12/29/2017] [Indexed: 10/18/2022]
|
17
|
Varjak M, Donald CL, Mottram TJ, Sreenu VB, Merits A, Maringer K, Schnettler E, Kohl A. Characterization of the Zika virus induced small RNA response in Aedes aegypti cells. PLoS Negl Trop Dis 2017; 11:e0006010. [PMID: 29040304 PMCID: PMC5667879 DOI: 10.1371/journal.pntd.0006010] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 11/02/2017] [Accepted: 10/04/2017] [Indexed: 01/16/2023] Open
Abstract
RNA interference (RNAi) controls arbovirus infections in mosquitoes. Two different RNAi pathways are involved in antiviral responses: the PIWI-interacting RNA (piRNA) and exogenous short interfering RNA (exo-siRNA) pathways, which are characterized by the production of virus-derived small RNAs of 25–29 and 21 nucleotides, respectively. The exo-siRNA pathway is considered to be the key mosquito antiviral response mechanism. In Aedes aegypti-derived cells, Zika virus (ZIKV)-specific siRNAs were produced and loaded into the exo-siRNA pathway effector protein Argonaute 2 (Ago2); although the knockdown of Ago2 did not enhance virus replication. Enhanced ZIKV replication was observed in a Dcr2-knockout cell line suggesting that the exo-siRNA pathway is implicated in the antiviral response. Although ZIKV-specific piRNA-sized small RNAs were detected, these lacked the characteristic piRNA ping-pong signature motif and were bound to Ago3 but not Piwi5 or Piwi6. Silencing of PIWI proteins indicated that the knockdown of Ago3, Piwi5 or Piwi6 did not enhance ZIKV replication and only Piwi4 displayed antiviral activity. We also report that the expression of ZIKV capsid (C) protein amplified the replication of a reporter alphavirus; although, unlike yellow fever virus C protein, it does not inhibit the exo-siRNA pathway. Our findings elucidate ZIKV-mosquito RNAi interactions that are important for understanding its spread. The recent outbreak of Zika virus (ZIKV) in the Americas has resulted in a severe threat to public health. ZIKV is transmitted by Aedes aegypti mosquitoes, thus it is important to understand virus-vector interactions. Analysis of ZIKV infection in mosquito cells indicated that two RNA interference pathways are involved during infection: the exogenous short-interfering (si)RNA (exo-siRNA) and PIWI-interacting (pi)RNA pathways. If Dcr2, an enzyme responsible for cleaving dsRNA into siRNAs, is knocked out, ZIKV replication is increased compared to control cells. However, the knockdown of Ago2 expression had no significant enhancing effect on ZIKV replication. In the case of the PIWI pathway, only the Piwi4 protein was found to have significant antiviral activity. Furthermore, unlike the capsid (C) protein of yellow fever virus, ZIKV capsid protein does not suppress the siRNA pathway. These results suggest that ZIKV has mechanisms to evade mosquito innate immunity and it is therefore important to understand these virus-vector interactions and the implications they have on transmission.
Collapse
Affiliation(s)
- Margus Varjak
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
- * E-mail: (MV); (AK)
| | - Claire L. Donald
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Timothy J. Mottram
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Vattipally B. Sreenu
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Andres Merits
- Institute of Technology, University of Tartu, Nooruse 1, Tartu, Estonia
| | - Kevin Maringer
- Department of Microbial Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Esther Schnettler
- Bernhard-Nocht-Institute for Tropical Medicine, Bernhard-Nocht-Strasse, Hamburg, Germany
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
- * E-mail: (MV); (AK)
| |
Collapse
|